US7485452B2 - Method of making optically active ester derivatives and their acids from racemic esters - Google Patents

Method of making optically active ester derivatives and their acids from racemic esters Download PDF

Info

Publication number
US7485452B2
US7485452B2 US11/587,228 US58722806A US7485452B2 US 7485452 B2 US7485452 B2 US 7485452B2 US 58722806 A US58722806 A US 58722806A US 7485452 B2 US7485452 B2 US 7485452B2
Authority
US
United States
Prior art keywords
hydroxybutyrate
ethyl
ester derivatives
optically active
racemic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US11/587,228
Other versions
US20080038802A1 (en
Inventor
Soon Ook Hwang
Sun Ho Chung
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Enzytech Ltd
Original Assignee
Enzytech Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Enzytech Ltd filed Critical Enzytech Ltd
Assigned to ENZYTECH, LTD. reassignment ENZYTECH, LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHUNG, SUN HO, HWANG, SOON OOK
Publication of US20080038802A1 publication Critical patent/US20080038802A1/en
Application granted granted Critical
Publication of US7485452B2 publication Critical patent/US7485452B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B6/00Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings
    • G02B6/0001Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings specially adapted for lighting devices or systems
    • G02B6/0011Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings specially adapted for lighting devices or systems the light guides being planar or of plate-like form
    • G02B6/0033Means for improving the coupling-out of light from the light guide
    • G02B6/005Means for improving the coupling-out of light from the light guide provided by one optical element, or plurality thereof, placed on the light output side of the light guide
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P41/00Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture
    • C12P41/003Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture by ester formation, lactone formation or the inverse reactions
    • C12P41/005Processes using enzymes or microorganisms to separate optical isomers from a racemic mixture by ester formation, lactone formation or the inverse reactions by esterification of carboxylic acid groups in the enantiomers or the inverse reaction
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P13/00Preparation of nitrogen-containing organic compounds
    • C12P13/001Amines; Imines
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P13/00Preparation of nitrogen-containing organic compounds
    • C12P13/002Nitriles (-CN)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P7/00Preparation of oxygen-containing organic compounds
    • C12P7/62Carboxylic acid esters
    • GPHYSICS
    • G02OPTICS
    • G02BOPTICAL ELEMENTS, SYSTEMS OR APPARATUS
    • G02B6/00Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings
    • G02B6/0001Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings specially adapted for lighting devices or systems
    • G02B6/0011Light guides; Structural details of arrangements comprising light guides and other optical elements, e.g. couplings specially adapted for lighting devices or systems the light guides being planar or of plate-like form
    • G02B6/0033Means for improving the coupling-out of light from the light guide
    • G02B6/0058Means for improving the coupling-out of light from the light guide varying in density, size, shape or depth along the light guide
    • G02B6/0061Means for improving the coupling-out of light from the light guide varying in density, size, shape or depth along the light guide to provide homogeneous light output intensity
    • GPHYSICS
    • G02OPTICS
    • G02FOPTICAL DEVICES OR ARRANGEMENTS FOR THE CONTROL OF LIGHT BY MODIFICATION OF THE OPTICAL PROPERTIES OF THE MEDIA OF THE ELEMENTS INVOLVED THEREIN; NON-LINEAR OPTICS; FREQUENCY-CHANGING OF LIGHT; OPTICAL LOGIC ELEMENTS; OPTICAL ANALOGUE/DIGITAL CONVERTERS
    • G02F1/00Devices or arrangements for the control of the intensity, colour, phase, polarisation or direction of light arriving from an independent light source, e.g. switching, gating or modulating; Non-linear optics
    • G02F1/01Devices or arrangements for the control of the intensity, colour, phase, polarisation or direction of light arriving from an independent light source, e.g. switching, gating or modulating; Non-linear optics for the control of the intensity, phase, polarisation or colour 
    • G02F1/13Devices or arrangements for the control of the intensity, colour, phase, polarisation or direction of light arriving from an independent light source, e.g. switching, gating or modulating; Non-linear optics for the control of the intensity, phase, polarisation or colour  based on liquid crystals, e.g. single liquid crystal display cells
    • G02F1/133Constructional arrangements; Operation of liquid crystal cells; Circuit arrangements
    • G02F1/1333Constructional arrangements; Manufacturing methods
    • G02F1/133342Constructional arrangements; Manufacturing methods for double-sided displays

Definitions

  • the present invention relates to a process for the preparation of optically active ⁇ -hydroxybutyl ester derivatives and their acid derivatives.
  • this invention relates to the process for preparing optically active ⁇ -hydroxybutyl ester derivatives and their acid derivatives by the hydrolysis of racemic ⁇ -hydroxybutyl ester derivatives represented by the general formula 1 in scheme 1 using lipases or lipase-producing microorganisms.
  • optically active ⁇ -hydroxybutyl ester derivatives and their acid derivatives can be used intensively as important chiral intermediates.
  • ⁇ -hydroxybutyl ester derivatives and their acid derivatives produced by this invention have high optical purity and this method can be used in practical process because separation and recovery of the products are easy. Therefore this invention can be used on the industrial scale.
  • ethyl (R)-3-hydroxybutyrate is an intermediate for an anti-glaucoma drug (Chirality in industry ⁇ . Chichester, UK:Wiley, 1997, 245-262) and (S)-3-hydroxybutyrate is used for synthesizing pheromones (Tertahedron, 1989, 45:3233-3298) and carbapenems (Journal of the Chemical Society. Perkin Transaction, 1999, 1: 2489-2494).
  • ethyl (R)-4-chloro-3-hydroxybutyrate is used for synthesizing L-carnitine(Journal of the American Chemical Society, 1983, 105:5925-5926), (R)-4-amino-3-hydroxybutyric acid (GABOB) and (R)-hydroxy-2-pyrrolidone.
  • Ethyl (S)-4-chloro-3-hydroxybutyrate is a valuable synthon for the production of hydroxyymethylglutaryl CoA (HMG-CoA) reductase inhibitor (Journal of Medicinal Chemistry, 1990, 33:2952-2956).
  • Ethyl (S)-3-hydroxybutyrate is synthesized by asymmetric hydrogenation of ethylacetoacetate using BINAP-coordinated Ru(II) complexes (Journal of the American Chemical Society, 1987, 109:5856-5858).
  • this method has disadvantages of high pressure during the reaction and high cost of metal catalyst.
  • ethyl (R)-3-hydroxybutyrate can be prepared by acidic alcoholysis of Poly-(R)-3-hydroxybutyrate accumulated by microorganisms (Enzyme and Microbial Technology, 2000, 27:33-36).
  • Optically active ethyl 4-chloro-3-hydroxybutyrate can be produced by reduction of ethyl 4-chloroacetoacetate.
  • Matsuyama et al. Japan Kokai Tokkyo Koho, 06-209782, Aug. 2, 1994
  • ethyl (S)-4-chloro-3-hydroxybutyrate 97% yield, 98% e.e) using Kluyveromyces lactis NRIC 1329.
  • Yamamoto et al. (Bioscience Biotechnology and Biochemistry, 2002, 66(2):481-483) produced ethyl (R)-4-chloro-3-hydroxybutyrate (95.2% conversion, 99% e.e) using recombinant microorganism expressing secondary alcohol dehydrogenase from Candida parapsilosis .
  • these methods have disadvantage of long reaction time.
  • Hoff et al. (Tetrahedron:Asymmetry, 1999, 10:1401-1412) obtained ethyl (S)-4-chlro-3-hydroxybutyrate (24% yield, 86% e.e) by transesterifying for 5 days using Rhizomucor miehei lipase (RML) in organic phase (benzene).
  • RML Rhizomucor miehei lipase
  • Suzuki et al. (Enzyme Microbiology and Technology, 1999, 24:13-20) produced ethyl (R)-4-chloro-3-hydroxybutyrate (99.8% e.e) using dechlorinase-producing microorganism.
  • optically active ⁇ -hydroxybutyl ester derivatives can be prepared by the stereoselective reduction of keto esters or the enzymatic transesterification.
  • these methods are not suitable due to their disadvantages including low enantiomeric excess, low yield or difficulties in the separation of products and reactants after reaction.
  • hydrolysis of ⁇ -hydroxybutyl ester derivatives Santaniello et al.(Gazzetta Chemica Italiana, 1989, 119:581-584) obtained ethyl (R)-4-chloro-3-hydroxybutyrate and (S)-their acid by hyrolysis of ethyl 4-chloro-3-hydroxybutyrate using pig liver esterase.
  • this method has disadvantages of low yield (23%) and low enantiomeric excess (16% e.e) and is not suitable for industrial use.
  • This method is simple and ester derivatives and their acid derivatives of higher optical purity can be obtained comparing to the conventional methods.
  • the objective of this invention is to provide the method of preparing optically active esters and their acids from racemic ⁇ -hydroxybutyl ester derivatives using enzymes or microorganisms.
  • the present invention consists of the process for preparing high optically active ⁇ -hydroxybutyl ester derivatives and their acids from racemic ⁇ -hydroxybutyl ester derivatives by stereospecific hydrolysis using lipases or lipase-producing microorganisms as biocatalysts in aqueous phase or organic phase including aqueous solvent.
  • this invention relates to the process for preparing optically active ⁇ -hydroxybutyl ester derivatives and their acid derivatives by stereospecific hydrolysis of racemic ⁇ -hydroxybutyl ester derivatives using lipases or lipase-producing microorganisms as biocatalysts in aqueous phase or organic phase including aqueous solvent.
  • methyl 3-hydroxybutyrate, ethyl 3-hydroxybutyrate, butyl 3-hydroxybutyrate, ethyl 3-azido-3-hydroxybutyrate, ethyl 4-chloro-3-hydroxybutyrate, ethyl 4-bromo-3-hydroxybutyrate and ethyl 4-cyano-3-hydroxybutyrate are used as racemic ⁇ -hydroxybutyl esters, but the reactant is not restricted to them.
  • Non-limiting examples of the commercially available lipases include PS lipase from Amano Inc., Candida rugosa lipase and Novozyme 435 and non-limiting examples of the lipase-producing microorganism include Candida rugosa and Rhodococcus butanica.
  • optically active esters and their acids are separated respectively by solvent extraction method or column chromatography.
  • racemic compounds were determined by gas chromatography (Donam Instrument Inc. Model 6200) equipped with HP-FFAP (Agilent, Inc., 30 mm ⁇ 0.53 m) column. The oven temperature was maintained initially at 70° C. for 5 min and then raised at the rate of 10° C./min to 220° C., and maintained for 10 minutes. Helium gas is used as carrier at the rate of 2 ml/min, and compounds were detected using FID detector.
  • the typical retention time of the components in this invention were as follows:
  • Racemic ethyl 4-bromo-3-hydroxybutyrate and racemic ethyl 4-cyano-3-hydroxybutyrate were analyzed using the same method used in the analysis of racemic methyl 3-hydroxybutyrate except that oven temperature was increased at 20° C./min. In this condition racemic ethyl 4-bromo-3-hydroxybutyrate and racemic ethyl 4-cyano-3-hydroxybutyrate are detected at 11.7 min and 14.07 min respectively.
  • Optically active methyl 3-hydroxybutyrate, ethyl 3-hydroxybutyrate, butyl 3-hydroxybutyrate and ethyl 4-azido-3-hydroxybutyrate were determined by HPLC (Waters, Inc., Model 1525) equipped with chial column OD-H (Daicel, 0.46 cm ⁇ 25 cm) using hexane and isopropyl alcohol mixture (90:10) as mobile phase.
  • the absorbance was 220 nm and flow rate was 0.7 ml/min.
  • the typical retention time of the components in this invention was as follows:
  • Optically active ethyl 4-cyano-3-hydroxybutyrate was determined by a gas chromatography(Donam Instrument Inc. Model 6200) equipped with chiral column G-TA(Astec, 30 mm ⁇ 0.32 m). The oven temperature was maintained initially at 100° C. for 5 min and then raised to 170° C. at the rate of 10° C./min, and maintained for 20 minutes. Helium gas was used as carrier gas and column head pressure was maintained at 10 psi, and compounds were detected using FID detector. In this condition, the typical retention time of ethyl (R)-4-cyano-3-hydroxybutyrate and ethyl (S)-4-cyano-3-hydroxybutyrate was 16.75 min and 16.53 min respectively.
  • Optically active ethyl 4-chloro-3-hydroxybutyrate was determined by a HPLC (Lab Alliance, Model 201) equipped with chial column OB-H (Daicel, 0.46 cm ⁇ 25 cm) using hexane and isopropyl alcohol mixture (95:5) as mobile phase. The flow rate was 0.7 ml/min and absorbance was 215 nm. The typical retention time of ethyl (R)-4-chloro-3-hydroxybutyrate and ethyl (S)-4-chloro-3-hydroxybutyrate was 14.42 min and 15.38 min, respectively.
  • Optically active ethyl 4-bromo-3-hydroxybutyrate was determined by a HPLC (Lab Alliance, Model 201) equipped with chial column AD-H(Daicel, 0.46 cm ⁇ 25 cm) using hexane and isopropyl alcohol mixture (90:10) as mobile phase. The flow rate was 0.7 ml/min and absorbance was 220 nm. In this condition, the typical retention time of ethyl (R)-4-bromo-3-hydroxybutyrate and ethyl (S)-4-bromo-3-hydroxybutyrate was 12.23 min and 11.24 min, respectively.
  • Racemic ethyl 3-hydroxybutyrate (1%, v/v) was added to the vial containing 5 ml potassium phosphate buffer (pH 8.0, 0.1 M) and Novozyme 435 (4%, w/v). The reaction was carried out at 30° C. for 2 hours. The reaction mixture was extracted with ethyl acetate and analyzed by above-mentioned method. Ethyl (S)-3-hydroxybutyrate (97% e.e) was obtained from organic solvent at 55% conversion. The aqueous solution was acidified with hydrochloric acid and extracted with organic solvent. After esterification, ethyl (R)-3-hydroxybutyrate with optical purity of 80.4% e.e was obtained.
  • Lipase-producing microorganisms were isolated by cultivating in a medium containing 1% tributyrin. Isolated strains assimilated tributyrin and made clear zone. The strains were grown in LB medium or GYP medium including glucose, and microorganisms were harvested by centrifuge and used as biocatalysts. The results are shown in Table 1.
  • ethyl 4-azido-3-hydroxybutyrate was used instead of ethyl 3-hydroxybutyrate used in Example 1. The reaction was carried out for 1 hour and ethyl (S)-4-azido-3-hydroxybutyrate (80.2% e.e) was obtained at 83.5% conversion.
  • ethyl 4-bromo-3-hydroxybutyrate 1%, w/v
  • novozyme 435 lipase was used as a biocatalyst.
  • ethyl (R)-4-bromo-3-hydroxybutyrate 99% e.e was obtained at 88.3% conversion.
  • ethyl 4-cyano-3-hydroxybutyrate 1%, w/v
  • novozyme 435 lipase was used as a biocatalyst.
  • ethyl (R)-4-cyano-3-hydroxybutyrate 99% e.e was obtained at 57.3% conversion.
  • optically active ⁇ -hydroxybutyl ester derivatives can be produced easily by hydrolysis of this invention. With appropriate lipases or microorganisms, ⁇ -hydroxybutyl ester derivatives of high optical purity can be produced. Also, it is easy to separate optically active esters from their acids after reaction. Therefore, this method is a useful process on the industrial scale.

Abstract

The present invention relates to process for the preparing of optically active ester derivatives and their acid derivatives which are used intensively as important chiral intermediates from racemic β-hydroxybutyl ester derivatives. In more detail, this invention relates to the process for preparing optically active β-hydroxybutyl ester derivatives and their acid derivatives by stereospecific hydrolysis of racemic β-hydroxybutyl ester derivatives using Upases or lipase-producing microorganisms in the aqueous phase or organic phase including aqueous solvent. The method of making optically active ester derivatives and their acid derivatives by hydrolysis of β-hydroxybutyl ester derivatives represented by the general formula 1 in scheme 1 is easier and more economical comparing to the conventional methods and the products have high optical purity. Also separation of ester derivatives from acid derivatives is easy after reaction. Thus this method is a useful process on the industrial scale.

Description

TECHNICAL FIELD
The present invention relates to a process for the preparation of optically active β-hydroxybutyl ester derivatives and their acid derivatives. In more detail, this invention relates to the process for preparing optically active α-hydroxybutyl ester derivatives and their acid derivatives by the hydrolysis of racemic α-hydroxybutyl ester derivatives represented by the general formula 1 in scheme 1 using lipases or lipase-producing microorganisms.
The above-mentioned optically active α-hydroxybutyl ester derivatives and their acid derivatives can be used intensively as important chiral intermediates. Also, α-hydroxybutyl ester derivatives and their acid derivatives produced by this invention have high optical purity and this method can be used in practical process because separation and recovery of the products are easy. Therefore this invention can be used on the industrial scale.
Figure US07485452-20090203-C00001
According to a report, ethyl (R)-3-hydroxybutyrate is an intermediate for an anti-glaucoma drug (Chirality in industry Π. Chichester, UK:Wiley, 1997, 245-262) and (S)-3-hydroxybutyrate is used for synthesizing pheromones (Tertahedron, 1989, 45:3233-3298) and carbapenems (Journal of the Chemical Society. Perkin Transaction, 1999, 1: 2489-2494).
And ethyl (R)-4-chloro-3-hydroxybutyrate is used for synthesizing L-carnitine(Journal of the American Chemical Society, 1983, 105:5925-5926), (R)-4-amino-3-hydroxybutyric acid (GABOB) and (R)-hydroxy-2-pyrrolidone. Ethyl (S)-4-chloro-3-hydroxybutyrate is a valuable synthon for the production of hydroxyymethylglutaryl CoA (HMG-CoA) reductase inhibitor (Journal of Medicinal Chemistry, 1990, 33:2952-2956).
BACKGROUND ART
There are several methods to prepare optically active β-hydroxybutyl ester derivatives. Ethyl (S)-3-hydroxybutyrate is synthesized by asymmetric hydrogenation of ethylacetoacetate using BINAP-coordinated Ru(II) complexes (Journal of the American Chemical Society, 1987, 109:5856-5858). However, this method has disadvantages of high pressure during the reaction and high cost of metal catalyst.
Another method is the reduction of 3-oxo-esters using microorganisms. Jayasinghe et al. (Tetrahedron Letters, 1993, 34:3949-3950) obtained ethyl (S)-3-hydroxybutyrate (58% yield, 94% e.e) by the reduction of ethyl acetoacetate using freeze-dried yeast in petroleum ether and Medson et al.(Tetrahedron:Asymmetry, 1997, 8:1049-1054) obtained ethyl (S)-3-hydroxybutyrate (yield 69%, 99% e.e) from ethyl acetoacetate by the reduction using yeast in organic solvent. Chin-Joe et al.(Biotechnology and Bioengineering, 2000, 69:370-376) obtained ethyl (S)-3-hydroxybutyrate (99% e.e) at 85% conversion by the reduction of ethyl acetoacetate using Baker's yeast. However, these methods have disadvantages of low yield and purification problem after reaction.
On the other hand, Sugai et al.(Agricultural and Biological Chemistry, 1989, 53:2009-2010) obtained ethyl (S)-3-hydroxybutyrate (99.4% e.e) by transesterification of racemic ethyl 3-hydroxybutyrate using vinyl butanoate as an acylating agent and porcine pancreatic lipase as a catalyst. Fishman et al.(Biotechnology and Bioengineering, 2001, 74:256-263) obtained ethyl (S)-3-hydroxybutyrate (40% yield, 99.4% e.e) using CALB (Candida antartica) lipaseand vinyl acetate as an acyl donor.
In another case, ethyl (R)-3-hydroxybutyrate can be prepared by acidic alcoholysis of Poly-(R)-3-hydroxybutyrate accumulated by microorganisms (Enzyme and Microbial Technology, 2000, 27:33-36).
Optically active ethyl 4-chloro-3-hydroxybutyrate can be produced by reduction of ethyl 4-chloroacetoacetate. Matsuyama et al.(Japan Kokai Tokkyo Koho, 06-209782, Aug. 2, 1994) obtained ethyl (S)-4-chloro-3-hydroxybutyrate (97% yield, 98% e.e) using Kluyveromyces lactis NRIC 1329. Kataoka et al.(Applied microbiology and Biotechnology, 1999, 51:486-490) obtained ethyl (R)-4-chloro-3-hydroxybutyrate (94% yield, 92% e.e) using recombinant microorganism, which coexpress both the alcohol reductase Igene from Sporobolomyces salmonicolor and the glucose dehydrogenase gene from Bacillus megaterium. Yamamoto et al.(Bioscience Biotechnology and Biochemistry, 2002, 66(2):481-483) produced ethyl (R)-4-chloro-3-hydroxybutyrate (95.2% conversion, 99% e.e) using recombinant microorganism expressing secondary alcohol dehydrogenase from Candida parapsilosis. However, these methods have disadvantage of long reaction time.
On the other hand, Hoff et al.(Tetrahedron:Asymmetry, 1999, 10:1401-1412) obtained ethyl (S)-4-chlro-3-hydroxybutyrate (24% yield, 86% e.e) by transesterifying for 5 days using Rhizomucor miehei lipase (RML) in organic phase (benzene).
Suzuki et al.(Enzyme Microbiology and Technology, 1999, 24:13-20) produced ethyl (R)-4-chloro-3-hydroxybutyrate (99.8% e.e) using dechlorinase-producing microorganism.
As previously stated, optically active β-hydroxybutyl ester derivatives can be prepared by the stereoselective reduction of keto esters or the enzymatic transesterification. However, these methods are not suitable due to their disadvantages including low enantiomeric excess, low yield or difficulties in the separation of products and reactants after reaction. For solving these problems, there is hydrolysis of α-hydroxybutyl ester derivatives. Santaniello et al.(Gazzetta Chemica Italiana, 1989, 119:581-584) obtained ethyl (R)-4-chloro-3-hydroxybutyrate and (S)-their acid by hyrolysis of ethyl 4-chloro-3-hydroxybutyrate using pig liver esterase. However, this method has disadvantages of low yield (23%) and low enantiomeric excess (16% e.e) and is not suitable for industrial use.
DISCLOSURE OF INVENTION
Technical Problem
The process for preparing of β-hydroxybutyl ester derivatives and their acid derivatives of high optical purity was developed from racemic β-hydroxybutyl ester derivatives represented by the general formula 1 in scheme 1 by stereospecific hydrolysis using lipases or lipase-producing microorganisms.
This method is simple and ester derivatives and their acid derivatives of higher optical purity can be obtained comparing to the conventional methods.
Accordingly, the objective of this invention is to provide the method of preparing optically active esters and their acids from racemic β-hydroxybutyl ester derivatives using enzymes or microorganisms.
For the above objectives, the present invention consists of the process for preparing high optically active β-hydroxybutyl ester derivatives and their acids from racemic β-hydroxybutyl ester derivatives by stereospecific hydrolysis using lipases or lipase-producing microorganisms as biocatalysts in aqueous phase or organic phase including aqueous solvent.
Technical Solution
This invention is explained in more detail as follows. As mentioned above, this invention relates to the process for preparing optically active β-hydroxybutyl ester derivatives and their acid derivatives by stereospecific hydrolysis of racemic β-hydroxybutyl ester derivatives using lipases or lipase-producing microorganisms as biocatalysts in aqueous phase or organic phase including aqueous solvent.
In this invention, methyl 3-hydroxybutyrate, ethyl 3-hydroxybutyrate, butyl 3-hydroxybutyrate, ethyl 3-azido-3-hydroxybutyrate, ethyl 4-chloro-3-hydroxybutyrate, ethyl 4-bromo-3-hydroxybutyrate and ethyl 4-cyano-3-hydroxybutyrate are used as racemic β-hydroxybutyl esters, but the reactant is not restricted to them. In the general formula 1 in scheme 1, X is H, CN, N3, F, Cl, Br or I and R is CnH2n+1 (n=1˜8).
Non-limiting examples of the commercially available lipases include PS lipase from Amano Inc., Candida rugosa lipase and Novozyme 435 and non-limiting examples of the lipase-producing microorganism include Candida rugosa and Rhodococcus butanica.
After reaction, optically active esters and their acids are separated respectively by solvent extraction method or column chromatography.
In this invention, racemic compounds were determined by gas chromatography (Donam Instrument Inc. Model 6200) equipped with HP-FFAP (Agilent, Inc., 30 mm×0.53 m) column. The oven temperature was maintained initially at 70° C. for 5 min and then raised at the rate of 10° C./min to 220° C., and maintained for 10 minutes. Helium gas is used as carrier at the rate of 2 ml/min, and compounds were detected using FID detector. The typical retention time of the components in this invention were as follows:
racemic methyl 3-hydroxybutyrate-15.48 min
racemic ethyl 3-hydroxybutyrate-14.32 min
racemic butyl 3-hydroxybutyrate-17.16 min
racemic ethyl 4-azido-3-hydroxybutyrate-22.50 min
racemic ethyl 4-chloro-3-hydroxybutyrate-20.31 min
Racemic ethyl 4-bromo-3-hydroxybutyrate and racemic ethyl 4-cyano-3-hydroxybutyrate were analyzed using the same method used in the analysis of racemic methyl 3-hydroxybutyrate except that oven temperature was increased at 20° C./min. In this condition racemic ethyl 4-bromo-3-hydroxybutyrate and racemic ethyl 4-cyano-3-hydroxybutyrate are detected at 11.7 min and 14.07 min respectively.
Optically active methyl 3-hydroxybutyrate, ethyl 3-hydroxybutyrate, butyl 3-hydroxybutyrate and ethyl 4-azido-3-hydroxybutyrate were determined by HPLC (Waters, Inc., Model 1525) equipped with chial column OD-H (Daicel, 0.46 cm×25 cm) using hexane and isopropyl alcohol mixture (90:10) as mobile phase. The absorbance was 220 nm and flow rate was 0.7 ml/min. The typical retention time of the components in this invention was as follows:
methyl (R)-3-hydroxybutyrate-10.28 min
methyl (S)-3-hydroxybutyrate-12.44 min
ethyl (R)-3-hydroxybutyrate-12.77 min
ethyl (S)-3-hydroxybutyrate-11.43 min
butyl (R)-3-hydroxybutyrate-9.4 min
butyl (S)-3-hydroxybutyrate-10.64 min
ethyl (R)-4-azido-3-hydroxybutyrate-8.7 min
ethyl (S)-4-azido-3-hydroxybutyrate-10.86 min
Optically active ethyl 4-cyano-3-hydroxybutyrate was determined by a gas chromatography(Donam Instrument Inc. Model 6200) equipped with chiral column G-TA(Astec, 30 mm×0.32 m). The oven temperature was maintained initially at 100° C. for 5 min and then raised to 170° C. at the rate of 10° C./min, and maintained for 20 minutes. Helium gas was used as carrier gas and column head pressure was maintained at 10 psi, and compounds were detected using FID detector. In this condition, the typical retention time of ethyl (R)-4-cyano-3-hydroxybutyrate and ethyl (S)-4-cyano-3-hydroxybutyrate was 16.75 min and 16.53 min respectively.
Optically active ethyl 4-chloro-3-hydroxybutyrate was determined by a HPLC (Lab Alliance, Model 201) equipped with chial column OB-H (Daicel, 0.46 cm×25 cm) using hexane and isopropyl alcohol mixture (95:5) as mobile phase. The flow rate was 0.7 ml/min and absorbance was 215 nm. The typical retention time of ethyl (R)-4-chloro-3-hydroxybutyrate and ethyl (S)-4-chloro-3-hydroxybutyrate was 14.42 min and 15.38 min, respectively.
Optically active ethyl 4-bromo-3-hydroxybutyrate was determined by a HPLC (Lab Alliance, Model 201) equipped with chial column AD-H(Daicel, 0.46 cm×25 cm) using hexane and isopropyl alcohol mixture (90:10) as mobile phase. The flow rate was 0.7 ml/min and absorbance was 220 nm. In this condition, the typical retention time of ethyl (R)-4-bromo-3-hydroxybutyrate and ethyl (S)-4-bromo-3-hydroxybutyrate was 12.23 min and 11.24 min, respectively.
And racemic compounds were confirmed by FT-NMR (Burker, Model DRX300 or JEOL, Model AR400) and the results are as follows:
ethyl 4-azido-3-hydroxybutyrate:
1H-NMR (CDCl3, 300 MHz) δ(ppm)=1.28 (t, 3H), 2.53 (m, 2H), 3.28 (d, 1H), 3.34 (m, 2H), 4.21 (q, 2H)
ethyl 4-chloro-3-hydroxybutyrate:
1H-NMR (CDCl3, 400 MHz) δ(ppm)=1.28 (t, 3H), 2.62 (d, 2H), 3.53 (br, 1H), 3.60 (d, 2H), 4.20 (q, 2H), 4.33 (m, 1H)
ethyl 4-bromo-3-hydroxybutyrate:
1H-NMR (CDCl3, 400 MHz) δ(ppm)=1.28 (t, 3H), 2.7 (m, 2H), 3.48 (dd, 1H), 3.51 (dd, 1H), 4.17 (q, 2H), 4.20 (m, 1H)
ethyl 4-cyano-3-hydroxybutyrate:
1H-NMR (CDCl3, 300 MHz) δ(ppm)=1.26 (t, 3H), 2.5˜2.7 (m, 4H), 4.18 (q, 2H), 4.32 (m, 1H)
A better understanding of the present invention may be obtained through the following examples which are set forth to illustrate, but are not to be construed as the limit of the present invention.
EXAMPLE 1
Racemic ethyl 3-hydroxybutyrate (1%, v/v) was added to the vial containing 5 ml potassium phosphate buffer (pH 8.0, 0.1 M) and Novozyme 435 (4%, w/v). The reaction was carried out at 30° C. for 2 hours. The reaction mixture was extracted with ethyl acetate and analyzed by above-mentioned method. Ethyl (S)-3-hydroxybutyrate (97% e.e) was obtained from organic solvent at 55% conversion. The aqueous solution was acidified with hydrochloric acid and extracted with organic solvent. After esterification, ethyl (R)-3-hydroxybutyrate with optical purity of 80.4% e.e was obtained.
EXAMPLES 2-3
Instead of lipase used in Example 1, whole cells (20% (w/v)) were used. Lipase-producing microorganisms were isolated by cultivating in a medium containing 1% tributyrin. Isolated strains assimilated tributyrin and made clear zone. The strains were grown in LB medium or GYP medium including glucose, and microorganisms were harvested by centrifuge and used as biocatalysts. The results are shown in Table 1.
TABLE 1
Con- % e.e
Reaction version for Con-
Example Microorgnism time(hr) (%) ester figuration
2 Candida rugosa 62 69.6 99 S
KCCM 50521
3 Rhodococcus 3.5 63.9 99 S
butanica ATCC
21197
EXAMPLES 4-5
Instead of ethyl 3-hydroxybutyrate used in Example 1, 1% methyl 3-hydroxybutyrate and 5% butyl 3-hydroxybutyrate were used as reactants. The reaction was carried out with novozyme 435 lipase and the results are shown in Table 2.
TABLE 2
Con- % e.e
Reaction version for Con-
Example Reactant time(hr) (%) ester figuration
4 methyl 3- 5 78.0 98.8 S
hydroxybutyrate
5 butyl 3- 6 82.0 78.5 S
hydroxybutyrate
EXAMPLE 6
Instead of ethyl 3-hydroxybutyrate used in Example 1, ethyl 4-azido-3-hydroxybutyrate was used. The reaction was carried out for 1 hour and ethyl (S)-4-azido-3-hydroxybutyrate (80.2% e.e) was obtained at 83.5% conversion.
EXAMPLES 7-8
Instead of ethyl 3-hydroxybutyrate used in Example 1, ethyl 4-chloro-3-hydroxybutyrate was used as a reactant and lipases were used as biocatalysts. The results are shown in Table 3.
TABLE 3
Con- % e.e
Reaction version for Con-
Example Lipase time(hr) (%) ester figuration
7 Pseudomonas 22 71.0 99 S
cepatia lipase
8 Candida rugosa 32 76.0 99 R
lipase
EXAMPLE 9
Instead of ethyl 3-hydroxybutyrate used in Example 1, ethyl 4-bromo-3-hydroxybutyrate (1%, w/v) was used as a reactant and novozyme 435 lipase was used as a biocatalyst. After reaction for 1 hour 40 minutes, ethyl (R)-4-bromo-3-hydroxybutyrate (99% e.e) was obtained at 88.3% conversion.
EXAMPLE 10
Instead of ethyl 3-hydroxybutyrate used in Example 1, ethyl 4-cyano-3-hydroxybutyrate (1%, w/v) was used as a reactant and novozyme 435 lipase was used as a biocatalyst. After reaction for 3 hours, ethyl (R)-4-cyano-3-hydroxybutyrate (99% e.e) was obtained at 57.3% conversion.
EXAMPLES 11-13
Instead of ethyl 3-hydroxybutyrate used in Example 2, ethyl 4-chloro-3-hydroxybutyrate was used as a reactant and microorganisms in Table 4 were used as biocatalysts. The results are shown in Table 4.
TABLE 4
Con- % e.e
Reaction version for Con-
Example Microorganism time (hr) (%) ester figuration
11 Candida rugosa 32 76.1 99 S
KCTC 7292
12 Candida rugosa 47 77.3 99 S
KCCM 50521
13 Rhodococcus 8 76.1 99 R
butanica ATCC
21197
INDUSTRIAL APPLICABILITY
In accordance with Examples 1-13, optically active β-hydroxybutyl ester derivatives can be produced easily by hydrolysis of this invention. With appropriate lipases or microorganisms, β-hydroxybutyl ester derivatives of high optical purity can be produced. Also, it is easy to separate optically active esters from their acids after reaction. Therefore, this method is a useful process on the industrial scale.

Claims (2)

1. A process for preparing optically active β-hydroxybutyl ester derivatives and their acids derivatives from a racemic β-hydroxybutyl ester derivative, the process comprising reacting a mixture of a racemic β-hydroxybutyl ester derivative of the formula (1)
Figure US07485452-20090203-C00002
and a lipase or lipase-producing microorganism as a biocatalyst, the reaction being carried out in the aqueous phase or organic phase including aqueous solvent and providing a β-hydroxybutyl ester derivative of the formula (2) and a β-hydroxybutyl acid derivative of the formula (3),
Figure US07485452-20090203-C00003
wherein R is CnH2n+1,(n=1˜8) and X is H, N3, CN, F, Cl, Br or I.
2. A process according to claim 1, wherein the reaction mixture comprises a lipase-producing microorganism selected from Candida rugosa KCTC 7292, Candida rugosa KCCM 5521, or Rhodococcus butanica ATCC 21197.
US11/587,228 2004-04-29 2005-04-27 Method of making optically active ester derivatives and their acids from racemic esters Expired - Fee Related US7485452B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
KR1020040029791A KR100657212B1 (en) 2004-04-29 2004-04-29 The method of making optically active ester derivatives and their acids from racemic esters
KR10-2004-0029791 2004-04-29
PCT/KR2005/001213 WO2005111227A1 (en) 2004-04-29 2005-04-27 The method of making optically active ester derivatives and their acids from racemic esters

Publications (2)

Publication Number Publication Date
US20080038802A1 US20080038802A1 (en) 2008-02-14
US7485452B2 true US7485452B2 (en) 2009-02-03

Family

ID=35394166

Family Applications (1)

Application Number Title Priority Date Filing Date
US11/587,228 Expired - Fee Related US7485452B2 (en) 2004-04-29 2005-04-27 Method of making optically active ester derivatives and their acids from racemic esters

Country Status (5)

Country Link
US (1) US7485452B2 (en)
EP (1) EP1740714A1 (en)
KR (1) KR100657212B1 (en)
CN (1) CN1946855A (en)
WO (1) WO2005111227A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9163202B2 (en) 2013-08-02 2015-10-20 Eastman Chemical Company Aqueous cleaning compositions including an alkyl 3-hydroxybutyrate
US9249378B2 (en) 2013-08-02 2016-02-02 Eastman Chemical Company Aqueous cleaning compositions having enhanced properties
US9255059B2 (en) 2013-08-02 2016-02-09 Eastman Chemical Company Method for producing an alkyl 3-hydroxybutyrate
US9388114B2 (en) 2013-08-02 2016-07-12 Eastman Chemical Company Compositions including an alkyl 3-hydroxybutyrate
US10508070B1 (en) * 2018-11-28 2019-12-17 Shanghai Shine High International Trade Co., Ltd. Method for preparing 3-hydroxybutyrate salts
WO2023278240A1 (en) 2021-06-28 2023-01-05 Phb Industrial S.A. Methods to produce therapeutic formulations comprising hydroxybutirate and hydroxyvalerate, therapeutic formulations and uses thereof

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL159719A0 (en) 2001-07-06 2004-06-20 Teva Pharma Process for the preparation of 7-amino syn 3,5-dihydroxy heptanoic acid derivatives via 6-cyano syn 3,5-dihydroxy hexanoic acid derivatives
CN1948499B (en) * 2006-05-26 2010-12-08 江南大学 Method of preparing (R) 4,4,4-trifluoro 3-hydroxy ethyl butyrate by biocatalytic reaction
KR100893763B1 (en) 2007-08-29 2009-04-20 한국화학연구원 Method for preparing optically active alkyl 3-hydroxybutanoate derivatives
CN102321690B (en) * 2011-07-27 2012-07-18 陆宏国 Preparation method of ester chiral amines
CN104313064A (en) * 2014-10-01 2015-01-28 青岛科技大学 Method for producing chiral bromophenyl methyl propionate by virtue of cell method
CN105543191B (en) * 2016-02-24 2019-06-21 中国科学院南海海洋研究所 A kind of esterase PHE21 and its encoding gene and application

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4957867A (en) 1986-05-29 1990-09-18 Sumitomo Chemical Company, Limited Production of cyclopentenones by enzyme resolution
US5108916A (en) 1989-06-05 1992-04-28 Rhone-Poulenc Rorer, S.A. Process for stereoselectively hydrolyzing, transesterifying or esterifying with immobilized isozyme of lipase from candida rugosa
KR20000059531A (en) 1999-03-04 2000-10-05 조생현 Method for manufacturing the pirdo-benz-oxizine for optical a ativity
EP1408107A2 (en) 2002-10-07 2004-04-14 Daiso Co., Ltd. Chlorohydrin and hydroxycarboxylic ester asymmetric hydrolase gene

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4957867A (en) 1986-05-29 1990-09-18 Sumitomo Chemical Company, Limited Production of cyclopentenones by enzyme resolution
US5108916A (en) 1989-06-05 1992-04-28 Rhone-Poulenc Rorer, S.A. Process for stereoselectively hydrolyzing, transesterifying or esterifying with immobilized isozyme of lipase from candida rugosa
KR20000059531A (en) 1999-03-04 2000-10-05 조생현 Method for manufacturing the pirdo-benz-oxizine for optical a ativity
EP1408107A2 (en) 2002-10-07 2004-04-14 Daiso Co., Ltd. Chlorohydrin and hydroxycarboxylic ester asymmetric hydrolase gene

Non-Patent Citations (16)

* Cited by examiner, † Cited by third party
Title
Boaz, Neil W., Enzymatic Hydrolysis of Ethyl 3-Hydroxy-3-phenylpropanoate: Observations on an Enzyme Active-Site Model, J. Org. Chem. 57, 1992, pp. 4289-4292.
Bucciarelli et al., Enantioselective lipase-catalyzed acetylation of beta-lactam precursors of carbapenem antibiotics, J. Chem. Soc., Perkin Trans. 1, 1999, pp. 2489-2494.
Chin-Joe et al., Hydrolytic Activity in Baker's Yeast Limits the Yield of Asymmetric 3-Oxo Ester Reduction, Biotechnology and Bioengineering, 2000, vol. 69, No. 4, pp. 370-376.
Fishman et al., A Two-Step Enzymatic Resolution Process for Large-Scale Production of (S)- and (R)-Ethyl-3-Hydroxybutyrate, Biotechnology and Bioengineering, 2001, vol. 74, No. 3, pp. 256-263.
Hoff et al., Lipase-catalyzed resolution of esters of 4-chloro-3-hydroxybutanoic acid: effects of the alkoxy group and solvent on the enantiomeric ratio, Tetrahedron: Asymmetry 10, 1999, pp. 1401-1412.
Jayasinghe et al., The Yeast Medicated Reduction of Ethyl acetoacetate in Petroleum Ether, Tetrahedron Letters, 1993, vol. 34, No. 24, pp. 3849-3850.
Kataoka et al., Stereoselective reduction of ethyl 4-chloro-3-oxobutanoate by Escherichia coli transformant cells coexpressing the aldehyde reductase and glucose dehydrogenase genes, Appl. Microbiol Biotechnol, 51, 1999, pp. 486-490.
Lee et al., Preparation of alkyl (R)-(-)-3-hydroxybutyrate by acidic alcoholysis of poly-(R)-(-)-3-hydroxybutyrate, Enzyme and Microbial Technology 27, 2000, pp. 33-36.
Medson et al., The stereoselective preparation of beta-hydroxy esters using a yeast reduction in an organic solvent, Tetrahedron: Asymmetry, 1997, vol. 8, No. 7 , pp. 1049-1054.
Noyori et al., Asymmetric Hydrogenation of beta-Keto Carboxylic Esters. A Practical, Purely Chemical Access to beta-Hydroxy Esters in High Enantiomeric Purity, J. Am. Chem. Soc., 1987, 109, pp. 5856-5858.
Sharma et al, Journal of Molecular Catalysis B: Enzymatic, Enantio-reversal in Candida rugosa Lipase-catalyzed Esterification of 3-Hydroxybutyric Acid, 2000, 10, pp. 531-534. *
Shiosaki et al., Phosphorus-Containing Inhibitors of HMG-CoA Reductase. 1. 4-[(2-Arylethyl)hydroxyphosphinyl]-3-hydroxybutanoic Acids: A New Class of Cell-Selective Inhibitors of Cholesterol Biosynthesis, J. Med. Chem. 1990, 33, pp. 2952-2956.
Sugai et al., Enzymatic Preparation of Ethyl (S)-3-Hydroxybutanoate with a High Enantiomeric Excess, Agric. Biol. Chem, 1989, 53 (7), pp. 2009-2010.
Suzuki et al., Dual production of highly pure methyl (R)-4-chloro-3-hydroxybutyrate and (S)-3-hydroxy-gamma-butyrolactone with Enterobacter Sp., Enzyme and Microbial Technology 24, 1999, pp. 13-20.
Yamamoto et al., Synthesis of Ethyl (R)-4-Chloro-3-hydroxybutanoate with Recombinant Escherichia coli Cells Expressing (S)-Specific Secondary Alcohol Dehydrogenase, Biosci. Biotechnol. Biochem., 2002, 66 (2), pp. 481-483.
Zhou et al., Stereochemical Control of Yeast Reductions. 1. Asymmetric Synthesis of L-Carnitine, J. Am. Chem. Soc., 1983, 105, pp. 5925-5926.

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9163202B2 (en) 2013-08-02 2015-10-20 Eastman Chemical Company Aqueous cleaning compositions including an alkyl 3-hydroxybutyrate
US9249378B2 (en) 2013-08-02 2016-02-02 Eastman Chemical Company Aqueous cleaning compositions having enhanced properties
US9255059B2 (en) 2013-08-02 2016-02-09 Eastman Chemical Company Method for producing an alkyl 3-hydroxybutyrate
US9255241B2 (en) 2013-08-02 2016-02-09 Eastman Chemical Company Aqueous cleaning compositions including an alkyl 3-hydroxybutyrate
US9388114B2 (en) 2013-08-02 2016-07-12 Eastman Chemical Company Compositions including an alkyl 3-hydroxybutyrate
US9506017B2 (en) 2013-08-02 2016-11-29 Eastman Chemical Company Aqueous cleaning compositions including an alkyl 3-hydroxybutyrate
US10508070B1 (en) * 2018-11-28 2019-12-17 Shanghai Shine High International Trade Co., Ltd. Method for preparing 3-hydroxybutyrate salts
WO2023278240A1 (en) 2021-06-28 2023-01-05 Phb Industrial S.A. Methods to produce therapeutic formulations comprising hydroxybutirate and hydroxyvalerate, therapeutic formulations and uses thereof

Also Published As

Publication number Publication date
KR100657212B1 (en) 2006-12-14
CN1946855A (en) 2007-04-11
US20080038802A1 (en) 2008-02-14
WO2005111227A1 (en) 2005-11-24
KR20050104481A (en) 2005-11-03
EP1740714A1 (en) 2007-01-10

Similar Documents

Publication Publication Date Title
US7485452B2 (en) Method of making optically active ester derivatives and their acids from racemic esters
US9970043B2 (en) Process for producing optically active 2-alkyl-1,1,3-trialkoxycarbonylpropane
JP3794702B2 (en) Enzymatic preparation of chiral-α-tertiary carboxylic esters
US20100248337A1 (en) PROCESS FOR THE PRODUCTION OF (s)-5-CHLORO-2-ISOPROPYLPENT-4-ENOIC ACID ESTERS
Jeon et al. Synthesis of alkyl (R)-lactates and alkyl (S, S)-O-lactyllactates by alcoholysis of rac-lactide using Novozym 435
US20030018048A1 (en) Optically pure paroxetine precursors
JP4042454B2 (en) Process for producing optically active 3-methylglutaric acid monoester
EP0451668B1 (en) Process for the production of optically active alkyl 3-aryl-3-hydroxypropionates
Romano et al. A new chemoenzymatic synthesis of D-cloprostenol
Yasohara et al. Enzymatically enantioselective hydrolysis of prochiral 1, 3-diacyloxyglycerol derivatives
JP3024299B2 (en) Optically active cyclopentene alcohols, production method thereof and use thereof
JP3732535B2 (en) Process for producing optically active α-methylalkanedicarboxylic acid-ω-monoester and its enantiomer diester
KR100758512B1 (en) The method of preparing optically active 3-hydroxy-3-phenylpropionic acids and optically active 3-acyloxy-3-phenylpropionic acid by enzymatic method
KR100748897B1 (en) The method of making optically active 3-hydroxybutyric acid and their esters by enzymatic method
JP2007117034A (en) Method for producing optically active nipecotic acid compound
US5986095A (en) Enantioselective preparation of halophenyl alcohols and acylates
JP4746019B2 (en) Optically active β-cyanoisobutyric acid and process for producing the same
JP2615768B2 (en) Optically active carboxylic acid derivative and method for producing the same
JP4565672B2 (en) Optically active β-cyanoisobutyric acid and process for producing the same
JP2003520018A (en) Method for producing optically active 1-amino-4- (hydroxylmethyl) -cyclopent-2-ene derivative
US20030109029A1 (en) Process for the preparation of enantiomerically pure tertiary ss-hydroxycarboxylic acids or their esters
JP2009263341A (en) Method of manufacturing optically active nipecotic acid ester derivative
JP2010505417A (en) (3) Specific hydrolysis of N-unprotected (R) -esters of (3) -amino-3-arylpropionic acid esters
JP2006014636A (en) Method for producing acyl derivative
JP2006061112A (en) Method for producing optically active 2-(cyclopentylmethyl)-malonic acid monoester

Legal Events

Date Code Title Description
AS Assignment

Owner name: ENZYTECH, LTD., KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HWANG, SOON OOK;CHUNG, SUN HO;REEL/FRAME:020142/0759

Effective date: 20061010

FPAY Fee payment

Year of fee payment: 4

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20170203